Papain, A Biotechnological Success Story.

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Trends in Food Science & Technology xxx (2017) xxx-xxx

Contents lists available at ScienceDirect

Trends in Food Science & Technology

F
journal homepage: www.elsevier.com

OO
Review

“New trends for a classical enzyme: Papain, a biotechnological success story in the
food industry”
Jesús Fernández-Lucasa, b, ∗, Daniel Castañedab, Daniel Hormigoa, ∗∗

PR
a
Applied Biotechnology Group, Department of Pharmacy and Biotechnology, School of Biomedical Sciences, Universidad Europea de Madrid, Urbanización El Bosque, Calle
Tajo s/n, 28670 Villaviciosa de Odón, Madrid, Spain
b
Grupo de Investigación en Desarrollo Agroindustrial Sostenible, Department of Agroindustrial Engineering, School of Environmental Sciences, Universidad de la Costa, Cra. 55
#58-66, Barranquilla, Colombia

ARTICLE INFO ABSTRACT

D
Article history: Background
Received 4 April 2017 In recent years, proteases have arisen as standard biocatalysts in many industrial processes in different
Received in revised form 19 August 2017
Accepted 23 August 2017
Available online xxx

Keywords:
TE
fields, such as pharmaceutical, medicine, detergent manufacturing and food science. Among them, papain is
undoubtedly one of the most frequently studied and widely used proteases in the food industry around the
world. However, the latest advances in recombinant papain expression systems, genetically engineered biocat-
alysts, new purification and isolation strategies, and enzymatic immobilization will enhance the development
of new applications of papain, as well as improve and optimize classical applications.
Cysteine proteases Scope and approach
Papain This review addresses not only the latest advances in classic applications, such as meat tenderization and
EC
Enzyme biotechnology protein hydrolysates, but also the most innovative applications in different industries such as food, animal
Biocatalysis feed, bioactive peptides production, water treatment, baking and brewing, among many others.
Immobilization
In addition, papain is a perfect example of a successful industrial enzyme that covers all the steps of the
Industrial processes
biocatalytic cycle that are necessary for the industrial implementation of any biocatalyst. This cycle includes
the production and extraction of the enzyme concerned (from natural or recombinant sources), functional and
structural characterization, genetic improvement, immobilization and, finally, industrial application. This re-
view describes the complete biocatalytic cycle of papain.
RR

Key findings and conclusions


Papain is clearly a case of industrial and commercial success over the last 40 years. The key to this success
has been continual biotechnological and process engineering innovation, which has opened up a new range
of possibilities for this exciting biocatalyst. However, further efforts are needed in protein engineering and
characterization of new mutants to reach the full potential of this enzyme.
© 2017.
CO

1. Introduction teases are important enzymes in living organisms, being involved in


many different biological processes.
Within the global industrial enzymes market, the food industry Protease enzymes are used in a large variety of applications,
enzymes sector is the major segment and is expected to grow from mainly in the detergent and pharmaceutical industries, followed by the
nearly $1.5 billion in 2016 to $1.9 billion in 2021 (BCC Research food industry. Since proteases represent more than 60% of the enzyme
Biotechnology Report, 2011. Enzymes in industrial applications: market share, with an expected CAGR (compound annual growth rate)
Global markets). of 5.3% from 2014 to 2019, they are the most important type of com-
UN

Proteases (also called peptidases or proteinases) are enzymes that mercialized enzymes in the world. Leading producers worldwide in-
hydrolyse the peptidic linkages in protein into shorter fragments (pep- clude Novo Industries, DSM, DuPont Industries, BASF, Genencor In-
tides) and eventually into their components, amino acids. Pro ternational, and Roche (Feijoo-Siota & Villa, 2011; Kumar, Singh,
Sangwan, & Gill, 2014).
According to the nature of the active site, proteases can be divided
into seven mechanistic classes: serine proteases, S (EC 3.4.21); cys-
teine proteases, C (EC 3.4.22); aspartic and glutamic proteases, D or

Corresponding author. Applied Biotechnology Group, Department of Pharmacy and E (EC 3.4.23); metalloproteases, M (EC 3.4.24); threonine proteases,
Biotechnology, School of Biomedical Sciences, Universidad Europea de Madrid,
Urbanización El Bosque, Calle Tajo s/n, 28670 Villaviciosa de Odón, Madrid, Spain.
T (EC 3.4.25); and asparagine peptide lyases, N (EC 4.3) (). Accord-
∗∗
Corresponding author. ing to the MEROPS database, cysteine proteases are divided into ten
Email addresses: jesus.fernandez2@universidadeuropea.es (J. Fernández-Lucas); clans, and most plant cysteine proteases belong to the C1 family, also
daniel.hormigo@universidadeuropea.es (D. Hormigo)

https://doi.org/10.1016/j.tifs.2017.08.017
0924-2244/© 2017.
2 Trends in Food Science & Technology xxx (2017) xxx-xxx

known as the papain family (papain-like proteases) Rawlings et al., combinant sources, operational conditions, genetic modifications to
2010). Papain-like proteases are found in most known organisms, such improve its functional characteristics, enzyme immobilization, and in-
as viruses, protozoa, plants, invertebrates, and vertebrates. Most of dustrial applications. It also describes the most recent trends in meat
the C1 family members are endopeptidases, but some of them dis- tenderization, dairy industry, production of protein hydrolysates and
play a wide variety of activities, including aminopeptidases, dipeptidyl bioactive peptides, food allergens removal, brewing and baking indus-

F
peptidases and enzymes with both exo- and endo-peptidase activities try, animal feed and water treatment, among other fields. Finally, we
(Feijoo-Siota & Villa, 2011; Novinec & Lenarčič, 2013; Rawlings et look at some industrial uses of papain not strictly focused on the food
al., 2010). industry, but which demonstrate the enormous versatility of this ex-

OO
Papain (EC 3.4.22.2), also called Papaya proteinase I (PPI), is a citing enzyme, such as certain biomedical approaches, antimicrobial
23.4 kDa, 212 residue cysteine endopeptidase belonging to subfamily food packaging, caries removal agent, or tooth-whitening additive in
C1A of papain-like proteases (Rawlings et al., 2010). The commer- toothpastes.
cial importance of papain is mainly due to its strong proteolytic activ-
ity against a broad range of protein substrates, and because it is active 2. Production and purification strategies
across a broad range of operational conditions. These interesting char-
acteristics have allowed papain to lead the proteases market, outselling 2.1. Plant derived papain

PR
other plant derived proteases, such as bromelain (Ananas comosus)
and ficin (Ficus carica), as well as fungal source proteases (Market Papain can be obtained from the latex of the papaya plant (Car-
Research Future, 2017. Global Meat Tenderizing Agents Market Re- ica papaya), which is a natural source of other endopeptidases, such
search Report- Forecast to 2023). Major producers of papain include as chymopapain (EC 3.4.22.6), caricain (EC 3.4.22.30) and glycyl
India, Sri Lanka, Democratic Republic of Congo, Zaire, Tanzania, endopeptidase (EC 3.4.22.25). In fact, papain is a minor constituent
Uganda, Mexico, Brazil and Argentina. As regards the global papain (5–8%) among the papaya endopeptidases (Feijoo-Siota & Villa,
market, the main importers are concentrated in Europe and USA, with 2011).
a market size of about 150–200 and 300–400 tons per year, respec- Purification of papain from papaya latex has traditionally been car-

D
tively. Furthermore, the Japanese market is relatively small (less than ried out using precipitation methods, reaching high yields of up to 53 g
50 tons per year). Some of the main limiting factors of the global and of crude enzyme per kg of latex. Although these methods have rou-
sustainable supply of papain are the high climatic dependence of pa- tinely been used in industry, they can only achieve up to 39% purity
paya crops, as well as the economical and political issues of some pro- of papain (Nitsawang, Hatti-Kaul, & Kanasawud, 2006). An alterna-
ducing countries (IDEA, 2000. Commercialisation Bulletin 13 Papain
Report). This fact makes the search for alternative sources of papain
a priority for producing companies. Fortunately, the latest advances in
recombinant papain expression may provide a solution to this prob-
TE tive and more efficient purification strategy involves the use of various
chromatographic techniques (ion exchange or affinity). In these types
of techniques the initial pre-processing of the latex is essential before
samples can be applied on a chromatography column. This processing
lem. consists in a sun- or spray-drying process after latex extraction from
This review presents papain as an industrial success paradigm in plant (Feijoo-Siota & Villa, 2011).
EC
Biocatalysis, where all the steps necessary for the industrial imple- Current techniques usually include aqueous two-phase systems
mentation of any enzyme have been effectively addressed (Fig. 1). (ATPS). These systems may be composed of two polymers in aque-
The review provides a detailed description of each step, including the ous solution, or one polymer and salt in aqueous solution. ATPS tech-
different strategies of isolation and purification from natural and re niques have shown great potential for downstream processing of pa-
pain and other proteases, since they allow clarification, concentra
RR
CO
UN

Fig. 1. Complete biocatalytic cycle of papain.


Trends in Food Science & Technology xxx (2017) xxx-xxx 3

tion and purification of the target product in a one-pot process Eukariotic expression systems have also been studied for produc-
(Nitsawang et al., 2006). In this respect, it is worth mentioning that tion of recombinant papain. Vernet et al. (1990) obtained 0.3 mg/L
polymer–salt–water systems have aroused greater interest in the indus- of soluble papain by using a baculovirus/insect expression system.
try, since they are cheaper and show less viscosity than polymer–poly- Higher concentrations of 1.7 mg/L have been reported by Ramjee,
mer–water systems. In this regard, polyethylene glycol–phosphate Petithory, McElver, Weber, and Kirsch (1996) using the yeast Sac-

F
based systems are the most employed (Rocha & Nerli, 2013). Recent charomyces cerevisiae as expression system. More recently, Werner,
studies have demonstrated that the use of alginate as macro-ligand in Hirth, Rupp, and Zibek (2015) described the expression of a
PEG based systems improves papain purification. In a recent study, codon-optimized His-tagged papain in Pichia pastoris. After purifi-

OO
Rocha et al. (2016) described a PEG-citrate buffer system able to pu- cation 463 mg/L enzyme can be obtained, matching the highest pro-
rify papain from latex with a 20% of recovery. They observed that duction reported to date using E. coli as host (400 mg/L) (Choudhury
by adding alginate (0.1% w/w) and CaCl2 they could recover up to et al., 2009). Moreover, this codon-optimized papain can be purified
72% of papain and recycle PEG for purification. This strategy is very in a single step from the culture medium, and it showed a 1.4 times
promising, since it is low cost, easy to scale up, accurate and environ- higher enzymatic activity towards the chromogenic peptide Z-pheny-
mentally friendly (Rocha et al., 2016). lalanine-arginine-paranitroanilide compared with a commercial pa-
In 2014, He et al. reported an efficient method of large scale pa- pain. In this study, the authors demonstrate that the expression of pa-

PR
pain purification from unclarified papaya juice feedstock in batch pain is better in P. pastoris than in E. coli system (Werner et al., 2015).
adsorption system. In this study, the authors employed a reversed It is worth noting that the traditional process of papain production
phase expanded bed adsorption chromatography (RP-EBAC) using a from papaya crops (up to 53 g of crude enzyme per kg of wet latex)
FastlineTM-10-EBAC column packed with AmberliteTM-XAD-7HP. takes about 11–13 months from the transplant of Carica papaya un-
This technique allowed the authors to purify papain in a single oper- til the fruit is ready to produce the latex (Kamphuis, Kalk, Swarte, &
ation with a purification factor of 7.04 and a purity of 75% (He, bin Drenth, 1984). Comparing these data with those obtained by Werner
Tuan Chik, & Chong, 2014). et al. (2015) in Pichia pastoris, it can be estimated that a small culture
After the purification process, crude papain usually has to be in a 1000-L batch fermenter could yield 463 g of papain in as little as

D
treated with reducing agents in order to protect the free cysteine thiol 2–3 days. Taking into account this enormous difference in yields as
groups from oxidation, preserving its protease activity. When needed, well as the advantages that recombinant production offers regarding
free thiol groups of papain can be regenerated by the addition of low purification processes, irrespective of political, economic and climatic
molecular mass thiols, such as cysteine or dithiothreitol. factors, in our opinion the production of recombinant papain will com-

2.2. Recombinant papain


TE pletely displace traditional production in the medium term.

3. Operational conditions

The high world demand of papain for industrial uses makes neces- 3.1. Functional characterization
sary the search for alternative sources from traditional extraction from
EC
the latex of C. papaya. Despite the high recovery of papain from Car- Regarding its enzymatic activity, papain has a broad-spectrum en-
ica papaya plant material (up to 53 g of crude enzyme per kg of wet la- dopeptidase activity over a pH range of 5–8 and optimal temperature
tex), this natural source presents several drawbacks. First, papain only of 65 °C (Polaina & MacCabe, 2007).
represents about 5–8% of total cysteine proteases in the latex, which The specificity of papain has been extensively examined using
entails an expensive and ponderous isolation strategy (Nitsawang et different substrates, such as milk, haemoglobin, collagen discs and
al., 2006). Furthermore, isolated papain is highly susceptible to oxi- labelled collagen products, synthetic peptides, gelatine (You,
RR

dation, so it must immediately be conserved at low temperatures and Regenstein, & Liu, 2010), casein (Morais, Silva, Oliveira, & Silvestre,
direct air contact must be avoided. Another major disadvantage is the 2004), chitosane (Pan, Zeng, Foua, Alain, & Li, 2016), rice bran
dependence of papaya crops on external factors, such as climate, soil, protein concentrates (Ahmadifard, Murueta, Abedian-Kenari,
pests, etc., which avoids a continuous supply of papain. Motamedzadegan, & Jamali, 2016), soybean proteins (Shutov et al.,
The expression of recombinant papain in different microorgan- 2013), potato protein isolate (Waglay & Karboune, 2016), im-
ism-based systems makes it possible to overcome these drawbacks munoglobulins including sheep IgG, rabbit IgG, chicken IgY and
while significantly increasing the papain production capacity. Papain fish IgM, bovine serum albumin (BSA), lipid transfer protein (LTP),
CO

is synthesized as a pre-pro-peptide with 345 amino acid residues. This and whey proteins (α-lactalbumin and β-lactoglobulin) (Chalabi,
precursor contains a signal peptide (residues 1 to 18), a spacer peptide Khademi, Yarani, & Mostafaie, 2014), among many others (Table 1).
(19–33) and the mature enzyme (134–345). The most abundant amino
acids in papain precursor protein are glycine (35), tyrosine (28) and Table 1
valine (24), while histidine (3) and methionine (5) are present in the Optimum pH and temperature for papain with different substrates.
lowest amounts.
Substrate pH T (°C) References
Papain precursor can be cloned and expressed in Escherichia coli,
UN

but recombinant enzyme forms inactive inclusion bodies. However, Meat 7–8 60–65 (Bekhit et al., 2014)
they can be renatured to yields about 3 mg of active papain/L (Taylor Gelatin 7.5 56.8 (You et al., 2010)
et al., 1992). In this regard, Choudhury, Roy, Chakrabarti, Biswas, Casein 7.5 37 (Morais et al., 2004)
Chitosan 4.55 44.42 (Pan et al., 2016)
and Dattagupta (2009) described an optimized purification and refold- Rice bran proteins concentrates 8 28 (Ahmadifard et al., 2016)
ing protocol, reaching 400 mg/L of a His-tagged fusion pro-papain Soybean (β-conglycin) 5.6 30 (Shutov et al., 2013)
from E. coli. However, several problems can arise during the refold- Potato protein isolate 8 50 (Waglay & Karboune, 2016)
ing process, such as autoproteolysis, oxidation of the active site and Sheep and rabbit IgG 7 37 (Chalabi et al., 2014)
Fish IgM 5.5–8.5 37 (Chalabi et al., 2014)
formation of incorrect disulfide bonds. LTP 7.2 37 (Chalabi et al., 2014)
4 Trends in Food Science & Technology xxx (2017) xxx-xxx

According to the model proposed by Schechter and Berger (1967), Ménard, 1994). Another important residue is Gln19, which precedes
papain interacts with at least seven residues of the substrate, four the Cys25 and is believed to help in forming an oxyanion hole by sta-
amino acid residues in N-terminal direction from the cleaved bond bilizing the tetrahedral intermediate. Asn175 forms a hydrogen bond to
(named P1-P4), and three in C-terminal direction (named P1′-P3′). His159 (showing a similar role to the aspartic acid in the catalytic triad
Authors also proposed seven corresponding sites on the enzyme (sites of serine proteases), but is not necessary for catalysis (Vernet et al.,

F
S1-S4 and S1′-S3′) which are responsible for the recognition and 1995). Finally, it is interesting to note that Trp177 is involved in the
cleavage of polypeptide (Novinec & Lenarčič, 2013; Schechter & generation of the nucleophilic character of Cys25/His159 ion pair (Fig.
Berger, 1967). However, three decades later, the structural analysis of 2).

OO
reported papain structures support this definition to some extent, and In order to highlight the substrate binding-mode, a three-dimen-
only a few sites were confirmed in papain-like proteases (sites S2, S1 sional scheme of all described binding interactions is shown using the
and S1′) (Turk, Gunčar, Podobnik, & Turk, 1998). crystal structure of papain complexed with CLIK148, a representative
In general, papain-like proteases have broad specificity and the cathepsin L-specific inhibitor (Fig. 4) (Tsuge et al., 1999). On one
major determinant is the residue in the P2 position of the substrate. In side, the hydrogen bond formed between Asn175 and His159 stabilizes
this sense, Papain specificity is controlled by the S2 site, a hydropho- the substrate-binding pocket and also orients the His159 imidazolium
bic core that accommodates the P2 side chain of the substrate. Due ring. On the other side, the sulfhydryl group of catalytic Cys25 is cova-

PR
to this, papain shows high specificity for amino acids with hydropho- lently bonded to the C2 atom of CLIK148. Apart from that, CLIK148
bic or aromatic side chains, such as Val, Phe and Tyr, at this position. is stabilized in the active site by a network of hydrogen bonds. A hy-
However, papain does not recognize Val in P1′ (Choe et al., 2006; drogen bond is formed between the nitrogen atom of both Cys25 and
Groves, Coulombe, Jenkins, & Cygler, 1998; ; Novinec & Lenarčič, Gln19 and the oxygen atom O1 of CLIK148. Besides, the Gly66 pep-
2013; Turk et al., 1998). tide nitrogen is hydrogen bonded to the O3 atom of CLIK148. Another
hydrogen bond is formed between an oxygen atom of Asp158 and the
3.2. Structural characterization nitrogen atom N5 of CLIK148. As shown in Figs. 2 and 4, a water
molecule is located in the active site, near the residue His159. In this

D
Despite the limited sequence homology, cysteine proteases dis- regard, three hydrogen bonds are formed from N7 atom of CLIK148,
play a conserved core structure, composed of two clearly differenti- carbonyl oxygen of Asp158 and N1 atom of His159 to the water mole-
ated interacting domains, an α-helix and a β-barrel-like that have been cule. Finally, several hydrophobic interactions among CLIK148 with
termed the L- and R-domains according to their position in the stan- Trp177 (responsible for the stacking of pyridine ring of the substrate)
dard orientation (Fig. 2) (Novinec & Lenarčič, 2013). Papain struc-
ture displays a globular protein with three disulfide bonds and two
catalytic L- and R-domains. The active site is located at the inter-
face of L- and R-domains in the form of a V-shaped cleft and is
TE and Val133 are shown.

4. Biocatalyst improvement

formed by a cysteine (Cys), a histidine (His), an asparagine (Asn) 4.1. Genetic modifications
and a glutamine residue (Gln), which are conserved in all papain-like
EC
proteases (Fig. 2). In the mature protein, Cys25 and His159 form an As mentioned above, several studies have addressed papain mu-
ion pair substrate-binding pocket stabilized by Asn175, which orients tations to increase expression levels. Other studies, however, have
the His159 imidazolium ring. The sulfhydryl group of Cys25 performs told us more about the catalytic mechanism of this enzyme, such as
a nucleophilic attack on the carbonyl carbon of a substrate's peptide that carried out by Gul et al. (2008), who demonstrated that Asp158
bond, leading to an unstable tetrahedral intermediate. This interme- does not affect to nucleophilic character in the Cys25-His159 dyad as
diate spontaneously collapses to regenerate the carbonyl group, lead- previously thought, whereas Trp177 showed a key role. Choudhury,
RR

ing to an acyl enzyme complex, which is hydrolysed into the free en- Biswas, Roy, and Dattagupta (2010) developed single (Lys174→Arg),
zyme and the N-terminal portion of the substrate (Fig. 3) (Cstorer & double (Lys174→Arg, Val32→Ser) and triple mutants of papain
CO
UN

Fig. 2. A) Three-dimensional structure of papain (PDB accession code 1PPN). The arrow indicates the situation of active site. B) Catalytic residues in the active site of papain. Cys25
and His159 are forming the catalytic ion pair.
Trends in Food Science & Technology xxx (2017) xxx-xxx 5

F
OO
PR
D
TE
EC
Fig. 3. Catalytic mechanism of papain.

(Lys174→Arg, Val32→Ser, Gly36→Ser), increasing the thermostabil-


ity of the biocatalyst. The triple mutant was the most thermostable, in-
creasing its half-life by 45 min at 60 °C and at 65 °C compared with
wild type papain. In addition, the triple mutant had a faster inactiva-
tion rate beyond Tmax, which is very interesting for industrial imple-
RR

mentation.
Recent studies have demonstrated that the specificity of papain can
be modified by replacing the residue Ile86 in its pro-peptide region.
These mutations (Ile86→Phe, Ile86→Leu and Ile86→Ala) can block the
specificity determining S2-subsite of the catalytic cleft of the protease
in its zymogen form and significantly improve the macroscopic ki-
CO

netic parameters (Km and kcat) of the enzyme (Dutta, Choudhury, Roy,
Dattagupta, & Biswas, 2016). Several of these studies and others are
summarized in Table 2.

5. Papain immobilization

Enzyme immobilization provides an excellent base for increasing


UN

enzyme stability and half-life, also simplifying downstream process-


ing. Thus, many different methods exist for enzymatic immobiliza-
tion (physical adsorption, entrapment, copolymerization or covalent
Fig. 4. Crystal structure of CLIK148 (cyan C atoms with heteroatom coloring) bound in
the active site of papain. The hydrogen bonding interactions between ligand and protein attachment, among others), and several natural and synthetic supports
are draw as black dashed lines. Water molecule is draw as red sphere. (For interpreta- have been assessed for their efficiency in the process. Since papain
tion of the references to colour in this figure legend, the reader is referred to the web has been utilized in a large number of industrial applications, numer-
version of this article.) ous efficient strategies on a variety of carriers have been described
in the current literature, such as starch gel (Sangeetha & Abraham,
2006), nitrilon fibre functionalized with amine groups (Li, Xing, &
Ding, 2007), cotton fabric (Xue, Nie, Zhu, Li, & Zhang, 2010) or
6 Trends in Food Science & Technology xxx (2017) xxx-xxx

Table 2
Mutant variants of papain.

Mutation Host Result References

Lys174→Arg Lys174→Arg/Val32→Ser E. coli Increase thermal stability (Choudhury et al., 2010)

F
Lys174→Arg/Val32→Ser,Gly36→Ser
Ile86→Phe E. coli Modification of substrate specificity (Dutta et al., 2016)
Ile86→Leu
Ile86→Ala

OO
Gln19→Glu Baculovirus-insect cell expression system and S. Elucidation of the role of Gln19 in the active (Ménard et al., 1991)
cerevisiae site (Ménard et al., 1995)
Gln19→His
Gln19→Asn/Ser21→Ala
Gln19→ Ala
Gln19→ Ser
Val133→Ala/Ser205→Glu Baculovirus-insect cell expression system a Modification of substrate specificity of S2 (Khouri et al., 1991)
Val133→Ala/Val157→Gln/Ser205→Glu
Gly19→Glu Baculovirus-insect cell expression system Nitrile hydratase activity into papain (Dufour, Storer, & Ménard,

PR
1995)

sepharose (Homaei, Sajedi, Sariri, Seyfzadeh, & Stevanato, 2010), be- 6.1. Meat tenderization
tween many others.
More recent papain immobilization strategies have been included: The use of exogenous proteases to improve meat tenderness has
generation of porous cross-linked enzyme aggregates (p-CLEAs) become an increasing focus of interest recently. It is a priority for the
(Wang et al., 2011); the use of iron oxide magnetic nanoparticles meat industry to be able to cover the increasing demand for guaran-

D
(g-Fe2O3/Fe3O4) activated with thiophene and triazole as enzyme link- teed tender meat and give added value to lower-grade meat cuts.
ers (Xin, Si, & Xing, 2010); encapsulation in biosilica matrix through Many approaches have been based on improving post-mortem ten-
a biomimetic silicification process induced by papain (Zhou, Wang, derness, such as mechanical tenderization, water content enhance-
Jiang, & Gao, 2013); magnetic gold nanocomposites modified with ment, and different enzymatic treatments (Pietrasik & Shand, 2011).
3-(mercaptopropyl) trimethoxy silane (MPTS) (Sahoo, Sahu,
Bhattacharya, Dhara, & Pramanik, 2013) (Fig. 5); and crosslinking
with a glutaraldehyde to poly (vinyl alcohol) PVA nanofibers prepared
by electrospinning (Moreno-Cortez et al., 2015).
TE Traditionally, meat is softened by autoproteolysis (mainly mediated
by cathepsins and capains), keeping it at 4 °C for 7–10 days.
Under optimal operating conditions (pH around 7–8 and tempera-
ture between 60 and 65 °C) papain is able to hydrolyse almost any pro-
tein present in muscle tissue, as well as tendons and ligaments, which
makes it a potent meat softener (Bekhit, Hopkins, Geesink, Bekhit, &
EC
6. Industrial applications of papain Franks, 2014).
There are numerous studies in which the meat softening effect of
Functional properties of papain have developed an increasing in- papain has been evaluated when enzyme is injected into the animal
terest in a wide range of industrial uses, mainly in meat tenderization, before being slaughtered, allowing a homogenous distribution of pa-
foods, feeds, brewing and the textile industry. Papain has also been pain in the meat of the animal. Today, this technique presents ethi-
described as an active additive in tooth-bleaching dentifrices and skin cal concerns and is not used, since the injection of active papain can
RR

products. This part of the review summarises the main applications of cause suffering and stress in animals. An alternative is the injection
this enzyme (Table 3). of inactive papain. For this purpose, papain is usually treated with hy
CO
UN

Fig. 5. Schematic representation of the immobilization of papain on magnetic gold nanoparticles.


Trends in Food Science & Technology xxx (2017) xxx-xxx 7

Table 3 Post-mortem application is generally acceptable for lower-grade


Industrial applications of papain. meat cuts. Papain is supplied commercially in powder and liquid
Application Mode of action References forms (e.g. PANOL®, LIQUIPANOL® T100), as well as combined
with other proteases such as bromelain (e.g., ENZECO® DUAL PRO-
Meat Hydrolysis of connective tissue and myofibrillar (Pietrasik & TEASE). Several commercial preparations may include other ingredi-

F
tenderization proteins. Shand, 2011)
(Bekhit et al.,
ents (salt, phosphates or flavour enhancers such as sodium glutamate),
2014) and they are available to be used directly by processors and house-
Production Hydrolysis of proteins to form peptides of varying (Polaina & holds.

OO
of protein sizes with multiple applications: nutritional MacCabe,
hydrolysates supplement, pharmaceutical ingredient, flavor 2007)
6.2. Dairy industry
enhancer, bioactive properties, etc. (Agyei &
Danquah,
2011) Due to their ability to hydrolyse the specific peptide bonds to gen-
(Liu et al., erate casein and macropeptides, proteases have been extensively used
2016) as biocatalysts in milk clotting processes in the dairy industry. One of
(Meinlschmidt
et al., 2016)
the most important uses of proteases is cheese manufacturing. Tradi-

PR
Dairy Production of semisoft cheese or cream cheese, (Mahajan & tionally, dried calf stomachs or dried enzyme extracts (animal rennet)
industry improve the meltability and stretchability of Chaudhari, have been employed for better controlled cheese making. Due to lim-
cheese 2014) (Hejazin ited availability of calf stomachs, it was necessary to find new pro-
& El-Qudah, teases with similar properties, in order to be used as biocatalysts in
2009)
(Abe et al., the industrial production of cheese. The potential of plant proteases in
2015) cheese making has been reported in this regard (Jacob, Jaros, & Rohm,
Baking Increase of protein solubility, and reduction of (Polaina & 2011).
industry allergenic protein content of cereals. MacCabe, Arlene, Prima Kristijarti, and Ardelia (2015) studied the effect of
2007)

D
(Kong et al.,
papain in the production of Cheddar cheese with different types of
2007) milk (cow, goat and soy), concluding that papain displayed highly pro-
(Li et al., teolytic activity on milk but with a very irregular clotting performance.
2016) These results suggest that papain is not a good biocatalyst for the pro-
Animal feed Estimation of protein degradation in ruminant
feed. Production of bioactive peptides
(Polaina &
MacCabe,
2007)
(Choi et al.,
2016)
TE duction of hard cheese. However, papain shows significant advantages
over other proteases, such as better accessibility, lower price, greater
availability in large quantities, and more resistance to extreme pH and
temperatures. In this respect, if clotting performance could be con-
(Mo et al., trolled, papain could be used as a rennet enzyme replacement for the
2016)
production of hard cheese.
EC
Brewing and Degradation of several insoluble protein (Polaina &
wine aggregates formed during and after beer MacCabe, On the contrary, Mahajan and Chaudhari (2014) report the use of
industry fermentation, wine stabilization agent. 2007) papain in the production of semisoft cheese or cream cheese. Hejazin
(Esti et al., and El-Qudah (2009) improved the meltability and stretchability of
2013) Nabulsi cheese by papain addition. Finally, an interesting article de-
Bioethanol Papain acts as a deflocculating agent avoiding (Silva et al.,
industry yeast flocculation during fermentation. 2015) scribes how Abe, Wu, Kim, Fujii, and Abe (2015) developed a new
Water Binding of heavy metals, such as mercury, due to (Metin & Alver, method for soymilk cream production using a simple three-step
RR

treatment presence of four sulfhydryl groups on papain 2016) process, including papain digestion, heat treatment and low-speed cen-
active site. trifugation.
Tooth Removal of stains, plaque, and food debris from (Münchow et
whitening the outer tooth surface. al., 2016)
(Chakravarthy 6.3. Production of protein hydrolysates
& Acharya,
2012) Protein hydrolysates are widely used in industry with many dif-
Biomedicine Treatment of burn wounds, esophageal (Anuar, Zahari, ferent applications: nutritional supplement, pharmaceutical ingredi-
CO

obstruction, caries removal, activity against Taib, &


gastrointestinal nematodes, tissue repairing of Rahman, ent, flavor enhancer, nitrogen source for growth media for microbial,
venous ulcers, antibacterial activity, etc. 2008) plant, and animal cell culture, in cosmetics and in beverages. Cur-
(Bussadori et rently, the industrial production of protein hydrolysates includes acid,
al., 2014) alkali, and enzyme hydrolysis, among which the latter-mentioned is
(Ribeiro et al.,
2015)
acquiring the most importance in the food and pharmaceutical indus-
(Morse et al., tries. The enzymatic process is milder and facilitates control of the
2016) degree of hydrolysis, which makes the process efficient and repro-
UN

ducible. The operational parameters for enzymatic hydrolysis have


been extensively studied, the most important being temperature, hy-
drolysis time, pH, and degree of hydrolysis (DH) (Agyei & Danquah,
drogen peroxide to oxidize the catalytic cysteine and then injected into 2011).
the animal. Once the animal is sacrificed, the anoxic conditions in- As a result of hydrolysis, the parent protein forms peptides of vary-
duce the reduction of catalytic cysteine and thus the reactivation of pa- ing sizes depending on the enzyme employed and the operational pa-
pain (Bekhit et al., 2014). However, this ante-mortem method presents rameters. The peptides with a positive effect on health are termed
some drawbacks, mainly due to the difficulty in predicting the level of bioactive peptides, and they can regulate several physiological func-
tenderization, since this depends on different physiological factors of tions. Many bioactive qualities of protein hydrolysates with papain
the animal. Some problems that may appear are related to differences
in texture as compared with high quality meat slices, over-tenderiza-
tion, undesired tastes or smells, or degradation of organs that may be
of commercial interest.
8 Trends in Food Science & Technology xxx (2017) xxx-xxx

have been widely studied, such as antitumor, antioxidant, antidiabetic, hydrated. Therefore, gluten should be hydrolysed to obtain more
inhibition of angiotensin–I converting enzyme, modulation of immune mouldable dough. The use of proteases such as papain improves the
system and mineral binding ability (Nesse, Nagalakshmi, Marimuthu, quality of the dough, preventing it from contracting, and enhancing its
& Singh, 2011). solubility, softness and rising during baking process (Kong, Zhou, &
Due to their high antioxidant properties, fish protein hydolysates Qian, 2007; Polaina & MacCabe, 2007).

F
(FPH) have aroused special interest (Elavarasan & Shamasundar,
2016; Gajanan, Elavarasan, & Shamasundar, 2016). An important as- 6.5. Brewing and wine industry
pect of industrial production of FPH is related to the concentration

OO
methods that can be employed, such as oven, freeze or spray dry- Papain is a very commonly used enzyme in the brewing industry,
ing. Among them, freeze drying is the preferred operation in industry, especially in the production of light and clear beers, due to their high
since it causes less damage to peptides. However, high capital and run- chill-proofing potential (Polaina & MacCabe, 2007). The treatment
ning costs have led to a search for alternate drying methods. Recently, of beer with papain (among other proteases) allows the degradation
Elavarasan and Shamasundar (2016) compared the effect of oven- and of several insoluble protein aggregates formed during and after beer
freeze-drying on the antioxidant properties of FPH with papain from fermentation, without any side effects on its organoleptic properties.
Cirrhinus mrigala, suggesting that oven drying may be advantageous. These high protein levels are responsible for several undesired effects

PR
Moreover, enzymatic conversion of fish frame waste to protein hy- in beer, such as hazes, high viscosity and excess foaming.
drolysate could be a solution for minimizing the pollution issues re- In addition, due to the broad substrate specificity displayed by pa-
lated to seafood processing operations, and a way to add value to pro- pain, this protease could find useful application in winemaking. In this
cessing by-products. respect, several reported studies have tested the effect of papain as a
Another recent application of papain for the production of an- wine stabilization agent. In a recent study, Esti, Benucci, Lombardelli,
tioxidant hydrolysates is the Chinese walnut (Juglans regia L.) hy- Liburdi, and Garzillo (2013) evaluated papain activity under wine-like
drolysates (Liu et al., 2016). The authors hydrolysed walnut proteins conditions, showing that this protease could be applied efficiently as a
with different proteases including papain. This study revealed that hy- biocatalyst in the wine industry.

D
drolysates obtained with papain showed a yield and purity of 16% and
81%, respectively, higher than others. Moreover, approximately 99% 6.6. Animal feed
of peptides had a molecular weight lower than 1500 Da. Furthermore,
the bioassay indicated that peptides obtained with papain exhibited Approximately 90% of food energy and nutrients for ruminants is
one of the highest antioxidant properties.

of soy protein isolates (SPI), while sensory, technical and functional


properties can be improved. It is worth mentioning that papain proved
TE
In addition, papain can be used to reduce the level of allergenicity
provided by low-cost forages. Therefore, quantification of soluble ni-
trogenous compounds in the rumen is essential to evaluate the quality
of feed. Adding proteases like papain to animal feed can increase pro-
tein assimilation and bio-availability, which translates into significant
to be the most suitable protease for improving functionality and sen- economic savings (Polaina & MacCabe, 2007).
sory characteristics, effectively reducing the molecular weight of SPI Along these lines, the Hong Kong Agriculture, Fisheries and Con-
EC
(Meinlschmidt, Sussmann, Schweiggert-Weisz, & Eisner, 2016). servation Department has employed papain to hydrolyse proteins of
Another interesting application of papain in the production of pro- soy meal for fish feed. In this study, they supplement traditional pel-
tein hydrolysates is reported by Damodaran (2007). In this work, the lets with soybean hydrolysates and use them for feed three different
author studied the inhibition of ice crystal growth in ice creams by em- species of marine fishes (Rhabdosargus sarba, Epinephelus bleekeri
ploying gelatine hydrolysates, previously digested with papain. These and Trachinotus blochii). After 340 days of treatment, they observed
results are very important, because understanding the molecular inter- more relative weight gain and better growth yield in fishes fed with
RR

actions responsible for ice crystal growth inhibition by peptides from papain treated pellets (Mo, Lau, Kwok, & Wong, 2016). Moreover,
gelatine hydrolysate would greatly facilitate the development of new Choi, Lam, Mo, and Wong (2016) used mixtures of papain and brome-
peptide cryoprotective agents with enhanced antifreeze activity. lain to hydrolyse food wastes employed to make fish feed (final com-
position of proteases at 1–2%). They observed that the use of feeds
6.4. Baking industry supplemented with these mixtures of proteases improved the growth,
lipid accumulation and immunity of grass carp (Ctenopharyngodon
Wheat is one of the most harvested cereals throughout the world. idella).
CO

Nevertheless, wheat contains proteins able to cause allergic reactions


in 0.4–1.3% of children and 0.2–0.9% of adults. 6.7. Other industrial applications
Wheat grain contains different allergenic proteins such as gliadins
(a, b, c and x), that conserve the allergenic epitope Due to papain's strong proteolytic activity, it has been employed as
Gln-Gln-Gln-Pro-Pro. Papain has been used to produce hypoaller- a versatile tool in the food industry, but it has many other application
genic flour suitable for allergic consumers, since it is able to recog- fields, such as biomedicine, dental or textile industry, among others.
nize this -Gln-Pro rich motif. Li, Yu, Goktepe, and Ahmedna (2016) Here we want to report some industrial uses of papain not strictly fo-
UN

described a complete removal of gliadins of wheat flour by a sequen- cused on the food industry, but which reveal the enormous versatility
tial enzymatic treatment with alcalase and papain. This treatment was and commercial potential of this enzyme.
found to be more effective in reducing allergenic protein content and Applications of papain in medicine over the last five years include
increasing solubility compared with other proteases (Li et al., 2016). the treatment of proteinaceous esophageal food impaction using pa-
Another issue related to gluten in the baking industry is that it pain and other proteolytic enzymes as an initial treatment for all pa-
turns insoluble and expands to form lattice-like structures when it is tients with esophageal obstruction (Morse, Wang, Donahue, Garrity,
& Allan, 2016); the treatment of mild to moderate acne with a fixed
combination of 0.1% hydroxypinacolone retinoate (synthetic ester of
9-cis-retinoic acid), 1% retinol in glycospheres and 2% papain in gly
Trends in Food Science & Technology xxx (2017) xxx-xxx 9

cospheres in aqueous gel (Veraldi et al., 2015); and tissue repairing of growth of the papain market, in turn favouring the market for other
venous ulcers employing low concentrated papain gels (Ribeiro et al., proteases such as bromelain or fungal proteases.
2015). The latest advances in recombinant papain expression systems, es-
Nowadays, the use of papain is increasingly common in the dental pecially yeast-based systems, will help overcome these difficulties and
industry, where the proteolytic and antibacterial properties of papain provide the market with a cheaper and more sustainable supply of pa-

F
seem to be an efficient and safe alternative for caries removal before pain. In addition, the new strategies of purification and isolation, ge-
restorative procedures are undertaken. netic modification and enzymatic immobilization will improve the de-
Papain has also been employed as a chemo-mechanical caries re- velopment of new applications of papain, especially in the food indus-

OO
moval agent (CMCR), first commercialized in 2003 as Papacarie, try, making some processes profitable, which is currently not the case
which is a mixture of papain, chloramine, toluidine blue and salts with traditional production methods.
in a thickening emulsion (Bussadori et al., 2014). Other similar Papain remains a very important product in the meat, brewing and
chemo-mechanical caries removing agents based on papain are dairy industry, where the challenge is to improve the operational prop-
Apacaries (Juntavee et al., 2014), papEdent (Subramaniam & erties of papain and to reduce its supply. Regarding the latest trends,
Gilhotra, 2011) and CarieCare (Venkataraghavan et al., 2013). most of the efforts are focused on the production of bioactive peptides
One the other hand, papain has extensively been described as an and functional foods that may have antioxidant, antitumor, or hypoal-

PR
active ingredient in dentifrices. There are numerous types of tooth- lergenic properties.
paste containing various enzymes in order to remove stains from teeth In this respect, further efforts are needed in the functional charac-
(Chakravarthy & Acharya, 2012). More recently, Münchow, Hamann, terization of recombinant of papain, as well as in the generation of new
Carvajal, Pinal, and Bottino (2016) also described the stain removal mutant variants of papain with improved properties. In this sense, di-
effect of papain- and bromelain-based dentifrices applied to enamel. rected evolution, molecular docking, molecular dynamics and genetic
Another interesting example is the use of papain in antimicro- engineering could be very important tools in order to improve activity
bial food packaging. Cynthya, Prabhawathi, and Mukesh (2014) im- and stability of the enzyme.
mobilized papain on polyurethane films to avoid microbial contam-

D
ination on cheese. Experimental results showed that this derivative Compliance with ethical standards
could be used as effective agent control and reduce the growth of
Staphylococcus aureus biofilm formation. In the same way, Manohar, Funding
Prabhawathi, Sivakumar, and Doble (2015) tested the antimicrobial
effect of immobilized papain derivatives against Acinetobacter spp.
and S. aureus.
Other interesting uses of papain include water treatment for re-
moval of heavy metals (Metin & Alver, 2016), or preventing yeast
TE This work was supported by grant SAN151610 from the Santander
Foundation.

Conflict of interest
flocculation in the bioethanol industry (Silva, Rosa, Carvalho, &
Oliva-Neto, 2015). The authors declare that they have no conflict of interest.
EC
7. Conclusions Ethical approval

Papain is undoubtedly one of the most frequently studied and This article does not contain any studies with human participants
widely used proteases in the industry worldwide. Despite the fact or animals conducted by any of the authors.
that papain can cause an allergic response in humans (Díez-Gómez,
RR

Quirce, Aragoneses, & Cuevas, 1998; Mansfield, Ting, Haverly, & Acknowledgements
Yoo, 1985), papain is generally recognized as safe (GRAS) as a di-
rect human food ingredient according to Code of Federal Regula- We thank Peter Bonney for his continued support and enthusiasm
tions of US Food and Drug Administration (U. S. Food and Drug for the project.
Administration, 2016). Moreover, according to the FAO/WHO food
standards, papain is a food additive that may be used in the foods un-
References
der the conditions of good manufacturing practices (GMP) as outlined
CO

in the Preamble of the Codex GSFA (FAO/WHO Codex Alimentarius Abe, N., Wu, C.Y., Kim, Y.K., Fujii, T., Abe, K., 2015. Development of an efficient
Commission, 2016). soymilk cream production method by papain digestion, heat treatment, and
Although one cysteine protease inhibitor has been predicted to be low-speed centrifugation. Bioscience, Biotechnology, and Biochemistry 79,
in the papaya genome (Paull et al., 2008) and such inhibitor is also 1890–1892.
Agyei, D., Danquah, M.K., 2011. Industrial-scale manufacturing of pharmaceuti-
suspected to be stored in the papaya latex (Azarkan, El Moussaoui, cal-grade bioactive peptides. Biotechnology Advances 29, 272–277.
Van Wuytswinkel, Dehon, & Looze, 2003), specific papain inhibitors, Ahmadifard, N., Murueta, J.H.C., Abedian-Kenari, A., Motamedzadegan, A., Jamali,
which might compromise enzyme activity in papaya latex extracts H., 2016. Comparison the effect of three commercial enzymes for enzymatic hy-
UN

with relevance to the food industry, have so far not been found. drolysis of two substrates (rice bran protein concentrate and soy-been protein) with
SDS-PAGE. Journal of Food Science and Technology 53, 1279–1284.
Papain has been used in the food industry for decades and today
Anuar, N.S., Zahari, S.S., Taib, I.A., Rahman, M.T., 2008. Effect of green and ripe
continues to dominate the world market for industrial proteases. This Carica papaya epicarp extracts on wound healing and during pregnancy. Food and
is mainly due to its better operational characteristics, such as tem- Chemical Toxicology 46, 2384–2389.
perature stability and half-life, in comparison with its main competi- Arlene, A., Prima Kristijarti, A., Ardelia, I., 2015. The effects of the types of milk
tors. However, growing market demand is outpacing supply of pa- (cow, goat, soya) and enzymes (rennet, papain, bromelain) toward cheddar cheese
production. Makara Journal of Technology 19, 31–37.
pain, which has boosted prices in recent years and is holding back the Azarkan, M., El Moussaoui, A., Van Wuytswinkel, D., Dehon, G., Looze, Y., 2003.
Fractionation and purification of the enzymes stored in the latex of Carica papaya.
Journal of Chromatography B 790, 229–238.
BCC Research Biotechnology Report, 2011. Enzymes in industrial applications:
Global markets.
10 Trends in Food Science & Technology xxx (2017) xxx-xxx

Bekhit, A.A., Hopkins, D.L., Geesink, G., Bekhit, A.A., Franks, P., 2014. Exogenous mutans: An in vitro study. International Journal of Clinical Pediatric Dentistry 7,
proteases for meat tenderization. Critical Reviews in Food Science and Nutrition 77–81.
54, 1012–1031. Kamphuis, I.G., Kalk, K.H., Swarte, M.B., Drenth, J., 1984. Structure of papain re-
Bussadori, S.K., de Godoy, C.H.L., Alfaya, T.A., Fernandes, K.P.S., Mesquita-Ferrari, fined at 1.65 A resolution. Journal of Molecular Biology 179, 233–256.
R.A., Motta, L.J., 2014. Chemo-mechanical caries removal with Papacarie (TM): Khouri, H.E., Vernet, T., Ménard, R., Parlati, F., Laflamme, P., Tessier, D.C., et al.,
Case series with 84 reports and 12 months of follow-up. The Journal of Contempo- 1991. Engineering of papain: Selective alteration of substrate specificity by site-di-

F
rary Dental Practice 15, 250–253. rected mutagenesis. Biochemistry 30, 8929–8936.
Chakravarthy, P.K., Acharya, S., 2012. Efficacy of extrinsic stain removal by novel Kong, X., Zhou, H., Qian, H., 2007. Enzymatic hydrolysis of wheat gluten by pro-
dentifrice containing papain and bromelain extracts. Journal of Young Pharmacists teases and properties of the resulting hydrolysates. Food Chemistry 102, 759–763.
4, 245–249. Kumar, V., Singh, D., Sangwan, P., Gill, P.K., 2014. Global market scenario of indus-

OO
Chalabi, M., Khademi, F., Yarani, R., Mostafaie, A., 2014. Proteolytic activities of ki- trial enzymes. In: Beniwal, V., Sharma, A.K. (Eds.), Industrial enzymes: Trends,
wifruit actinidin (Actinidia deliciosa cv. Hayward) on different fibrous and globu- scope and relevance. Nova Science Publishers, New York, pp. 173–196.
lar proteins: A comparative study of actinidin with papain. Applied Biochemistry Liu, M.C., Yang, S.J., Hong, D., Yang, J.P., Liu, M., Lin, Y., et al., 2016. A simple
and Biotechnology 172, 4025–4037. and convenient method for the preparation of antioxidant peptides from walnut
Choe, Y., Leonetti, F., Greenbaum, D.C., Lecaille, F., Bogyo, M., Brömme, D., et al., (Juglans regia L.) protein hydrolysates. Chemistry Central Journal 10, 1–11.
2006. Substrate profiling of cysteine proteases using a combinatorial peptide li- Li, F.Y., Xing, Y.J., Ding, X., 2007. Immobilization of papain on cotton fabric by
brary identifies functionally unique specificities. Journal of Biological Chemistry sol–gel method. Enzyme and Microbial Technology 40, 1692–1697.
281, 12824–12832. Li, Y., Yu, J., Goktepe, I., Ahmedna, M., 2016. The potential of papain and alcalase
Choi, W.M., Lam, C.L., Mo, W.Y., Wong, M.H., 2016. Upgrading food wastes by enzymes and process optimizations to reduce allergenic gliadins in wheat flour.

PR
means of bromelain and papain to enhance growth and immunity of grass carp Food Chemistry 196, 1338–1345.
(Ctenopharyngodon idella). Environmental Science and Pollution Research 23, Mahajan, R.T., Chaudhari, G.M., 2014. Plant latex as vegetable source for milk clot-
7186–7194. ting enzymes and their use in cheese preparation. International Journal 2,
Choudhury, D., Biswas, S., Roy, S., Dattagupta, J.K., 2010. Improving thermostability 1173–1181.
of papain through structure-based protein engineering. Protein Engineering, De- Manohar, C.M., Prabhawathi, V., Sivakumar, P.M., Doble, M., 2015. Design of a pa-
sign and Selection 23, 457–467. pain immobilized antimicrobial food package with curcumin as a crosslinker.
Choudhury, D., Roy, S., Chakrabarti, C., Biswas, S., Dattagupta, J.K., 2009. Produc- PLoS One 10, e0121665.
tion and recovery of recombinant propapain with high yield. Phytochemistry 70, Mansfield, L.E., Ting, S., Haverly, R.W., Yoo, T.J., 1985. The incidence and clinical
465–472. implications of hypersensitivity to papain in an allergic population, confirmed by
Cstorer, A., Ménard, R., 1994. Catalytic mechanism in papain family of cysteine pepti- blinded oral challenge. Annals of Allergy 55, 541–543.

D
dases. Methods in Enzymology 244, 486–500. Market Research Future, 2017. Global meat tenderizing agents market Research re-
Cynthya, M., Prabhawathi, V., Mukesh, D., 2014. Papain immobilized polyurethane port- Forecast to 2023. ID: MRFR/F-B & N/2125-HCRR. In: www.
film as antimicrobial food package. World Academy of Science, Engineering and marketresearchfuture.com.
Technology, International Journal of Biological, Biomolecular, Agricultural, Food Meinlschmidt, P., Sussmann, D., Schweiggert-Weisz, U., Eisner, P., 2016. Enzymatic
and Biotechnological Engineering 8, 1367–1370.
Damodaran, S., 2007. Inhibition of ice crystal growth in ice cream mix by gelatin hy-
drolysate. Journal of Agricultural and Food Chemistry 55, 10918–10923.
Díez-Gómez, M.L., Quirce, S., Aragoneses, E., Cuevas, M., 1998. Asthma caused by
Ficus benjamina latex: Evidence of cross-reactivity with fig fruit and papain. An-
TE treatment of soy protein isolates: Effects on the potential allergenicity, technofunc-
tionality, and sensory properties. Food Science & Nutrition 4, 11–23.
Ménard, R., Carriere, J., Laflamme, P., Plouffe, C., Khouri, H.E., Vernet, T., et al.,
1991. Contribution of the glutamine 19 side chain to transition-state stabilization in
the oxyanion hole of papain. Biochemistry 30, 8924–8928.
nals of Allergy, Asthma & Immunology 80, 24–30. Ménard, R., Plouffe, C., Laflamme, P., Vernet, T., Tessier, D.C., Thomas, D.Y., et al.,
Dufour, E., Storer, A.C., Ménard, R., 1995. Engineering nitrile hydratase activity into a 1995. Modification of the electrostatic environment is tolerated in the oxyanion
EC
cysteine protease by a single mutation. Biochemistry 34, 16382–16388. hole of the cysteine protease papain. Biochemistry 34, 464–471.
Dutta, S., Choudhury, D., Roy, S., Dattagupta, J.K., Biswas, S., 2016. Mutation in the Metin, , Alver, E., 2016. Fibrous polymer-grafted chitosan/clay composite beads as a
pro-peptide region of a cysteine protease leads to altered activity and specificity-a carrier for immobilization of papain and its usability for mercury elimination. Bio-
structural and biochemical approach. PLoS One 11, e0158024. process and Biosystems Engineering 39, 1137–1149.
Elavarasan, K., Shamasundar, B.A., 2016. Effect of oven drying and freeze drying on Mo, W.Y., Lau, R.S.S., Kwok, A.C.K., Wong, M.H., 2016. Use of soybean meal and
the antioxidant and functional properties of protein hydrolysates derived from papain to partially replace animal protein for culturing three marine fish species:
freshwater fish (Cirrhinus mrigala) using papain enzyme. Journal of Food Science Fish growth and water quality. Environmental Pollution 219, 815–820.
and Technology 53, 1303–1311. Morais, H.A., Silva, V.D., Oliveira, M.C., Silvestre, M.P., 2004. Characterization of
RR

Esti, M., Benucci, I., Lombardelli, C., Liburdi, K., Garzillo, A.M.V., 2013. Papain free aromatic amino acids and estimation of exposition rate of phenylalanine in ca-
from papaya (Carica papaya L.) fruit and latex: Preliminary characterization in al- sein hydrolysates by second derivative spectrophotometry. Acta Científica Vene-
coholic–acidic buffer for wine application. Food and Bioproducts Processing 91, zolana 55, 1–6.
595–598. Moreno-Cortez, I.E., Romero-García, J., González-González, V., García-Gutierrez,
FAO/WHO Codex Alimentarius Commission, 2016. GSFA online: Codex general D.I., Garza-Navarro, M.A., Cruz-Silva, R., 2015. Encapsulation and immobiliza-
standard for food additives (GSFA) online database. In: http://www.fao.org/ tion of papain in electrospun nanofibrous membranes of PVA cross-linked with
gsfaonline/index.html. glutaraldehyde vapor. Materials Science and Engineering, C 52, 306–314.
Feijoo-Siota, L., Villa, T.G., 2011. Native and biotechnologically engineered plant pro- Morse, C.R., Wang, H., Donahue, D.M., Garrity, J.M., Allan, J.S., 2016. Use of prote-
teases with industrial applications. Food and Bioprocess Technology 4, olytic enzymes in the treatment of proteinaceous esophageal food impaction. The
CO

1066–1088. Journal of Emergency Medicine 50, 183–186.


Gajanan, P.G., Elavarasan, K., Shamasundar, B.A., 2016. Bioactive and functional Münchow, E.A., Hamann, H.J., Carvajal, M.T., Pinal, R., Bottino, M.C., 2016. Stain
properties of protein hydrolysates from fish frame processing waste using plant removal effect of novel papain-and bromelain-containing gels applied to enamel.
proteases. Environmental Science and Pollution Research 23, 24901–24911. Clinical Oral Investigations 20, 2315–2320.
Groves, M.R., Coulombe, R., Jenkins, J., Cygler, M., 1998. Structural basis for speci- Nesse, K.O., Nagalakshmi, A.P., Marimuthu, P., Singh, M., 2011. Efficacy of a fish
ficity of papain-like cysteine protease proregions toward their cognate enzymes. protein hydrolysate in malnourished children. Indian Journal of Clinical Biochem-
Proteins: Structure, Function, and Bioinformatics 32, 504–514. istry 26, 360–365.
Gul, S., Hussain, S., Thomas, M.P., Resmini, M., Verma, C.S., Thomas, E.W., et al., Nitsawang, S., Hatti-Kaul, R., Kanasawud, P., 2006. Purification of papain from Car-
2008. Generation of nucleophilic character in the Cys25/His159 ion pair of papain ica papaya latex: Aqueous two-phase extraction versus two-step salt precipitation.
UN

involves Trp177 but not Asp158. Biochemistry 47, 2025–2035. Enzyme and Microbial Technology 39, 1103–1107.
He, Y., bin Tuan Chik, S.M.S., Chong, F.C., 2014. Purification of papain from unclari- Novinec, M., Lenarčič, B., 2013. Papain-like peptidases: Structure, function, and evo-
fied papaya juice using reversed phase expanded bed adsorption chromatography lution. Biomolecular Concepts 4, 287–308.
(RP-EBAC). Journal of Industrial and Engineering Chemistry 6, 4293–4297. Pan, A.D., Zeng, H.Y., Foua, G.B., Alain, C., Li, Y.Q., 2016. Enzymolysis of chitosan
Hejazin, R.K., El-Qudah, M., 2009. Effect of proteases on meltability and stretchabil- by papain and its kinetics. Carbohydrate Polymers 135, 199–206.
ity of Nabulsi cheese. American Journal of Agricultural and Biological Sciences 4, Paull, R.E., Irikura, B., Wu, P., Turano, H., Chen, N.J., Blas, A., et al., 2008. Fruit de-
173–178. velopment, ripening and quality related genes in the papaya genome. Tropical
Homaei, A.A., Sajedi, R.H., Sariri, R., Seyfzadeh, S., Stevanato, R., 2010. Cysteine en- Plant Biology 1, 246–277.
hances activity and stability of immobilized papain. Amino Acids 38, 937–942. Pietrasik, Z., Shand, P.J., 2011. Effects of moisture enhancement, enzyme treatment,
Jacob, M., Jaros, D., Rohm, H., 2011. Recent advances in milk clotting enzymes. Inter- and blade tenderization on the processing characteristics and tenderness of beef
national Journal of Dairy Technology 64, 14–33. semimembranosus steaks. Meat Science 88, 8–13.
Juntavee, A., Peerapattana, J., Ratanathongkam, A., Nualkaew, N., Chatchiwiwattana, Polaina, J., MacCabe, A.P., 2007. Industrial enzymes: Structure, function and applica-
S., Treesuwan, P., 2014. The antibacterial effects of apacaries gel on Streptococcus tions. Springer, Netherland.
Trends in Food Science & Technology xxx (2017) xxx-xxx 11

Ramjee, M.K., Petithory, J.R., McElver, J., Weber, S.C., Kirsch, J.F., 1996. A novel Turk, D., Gunčar, G., Podobnik, M., Turk, B., 1998. Revised definition of substrate
yeast expression/secretion system for the recombinant plant thiol endoprotease binding sites of papain-like cysteine proteases. Biological Chemistry 379,
propapain. Protein Engineering, Design and Selection 9, 1055–1061. 137–148.
Rawlings, N.D., Barrett, A.J., Bateman, A., 2010. MEROPS: The peptidase database. U. S. Food and Drug Admistration (FDA), 2016. Direct food substances affirmed as
Nucleic Acids Research 38, D227–D233. generally recognized as safe. CFR - Code of Federal Regulations Title 21 (3), 184.
Rawlings, N.D., Barrett, A.J., Bateman, A., 2011. Asparagine Peptide Lyases A sev- Uganda’s Investment in Developing Export Agriculture (IDEA), 2000. Commercialisa-

F
enth catalytic type of proteolytic enzymes. The Journal of Biological Chemistry tion bulletins 13 papain report.
286, 38321–38328. Venkataraghavan, K., Kush, A., Lakshminarayana, C.S., Diwakar, L., Ravikumar, P.,
Ribeiro, A.P.L., Oliveira, B.G.R.B.D., Soares, M.F., Barreto, B.M.F., Futuro, D.O., Patil, S., et al., 2013. Chemomechanical caries removal: A review and study of an
Castilho, S.R.D., 2015. Effectiveness of 2% and 4% papain gels in the healing of indigen-ously developed agent (carie care TM gel) in children. Journal of Interna-

OO
venous ulcers. Revista Da Escola De Enfermagem Da U S P 49, 394–400. tional Oral Health 5, 84–90.
Rocha, M.V., Di Giacomo, M., Beltramino, S., Loh, W., Romanini, D., Nerli, B.B., Veraldi, S., Barbareschi, M., Guanziroli, E., Bettoli, V., Minghetti, S., Capitanio, B., et
2016. A sustainable affinity partitioning process to recover papain from Carica pa- al., 2015. Treatment of mild to moderate acne with a fixed combination of hydrox-
paya latex using alginate as macro-ligand. Separation and Purification Technology ypinacolone retinoate, retinol glycospheres and papain glycospheres. Giornale Ital-
168, 168–176. iano Di Dermatologia E Venereologia 150, 143–147.
Rocha, M.V., Nerli, B.B., 2013. Molecular features determining different partitioning Vernet, T., Tessier, D.C., Chatellier, J., Plouffe, C., Lee, T.S., Thomas, D.Y., et al.,
patterns of papain and bromelain in aqueous two-phase systems. International 1995. Structural and functional roles of asparagine 175 in the cysteine protease pa-
Journal of Biological Macromolecules 61, 204–211. pain. The Journal of Biological Chemistry 270, 16645–16652.
Sahoo, B., Sahu, S.K., Bhattacharya, D., Dhara, D., Pramanik, P., 2013. A novel ap- Vernet, T., Tessier, D.C., Richardson, C., Laliberté, F., Khouri, H.E., Bell, A.W., et al.,

PR
proach for efficient immobilization and stabilization of papain on magnetic gold 1990. Secretion of functional papain precursor from insect cells. Requirement for
nanocomposites. Colloids and Surfaces B: Biointerfaces 101, 280–289. N-glycosylation of the pro-region. The Journal of Biological Chemistry 265,
Sangeetha, K., Abraham, T.E., 2006. Chemical modification of papain for use in alka- 16661–16666.
line medium. Journal of Molecular Catalysis B: Enzymatic 38, 171–177. Waglay, A., Karboune, S., 2016. Enzymatic generation of peptides from potato pro-
Schechter, I., Berger, A., 1967. On the size of the active site in proteases. I. Papain. teins by selected proteases and characterization of their structural properties.
Biochemical and Biophysical Research Communications 27, 157–162. Biotechnology Progress 32, 420–429.
Shutov, A.D., Rudakova, A.S., Rudakov, S.V., Kakhovskaya, I.A., Schallau, A.A., Wang, M., Jia, C., Qi, W., Yu, Q., Peng, X., Su, R., et al., 2011. Porous-CLEAs of pa-
Wilson, K.A., et al., 2013. Degradation of β-conglycinin β-homotrimer by Papain: pain: Application to enzymatic hydrolysis of macromolecules. Bioresource Tech-
Independent occurrence of limited and extensive proteolysis. Bioscience, Biotech- nology 102, 3541–3545.
nology, and Biochemistry 77, 2082–2086. Werner, N., Hirth, T., Rupp, S., Zibek, S., 2015. Expression of a codon-optimized Car-

D
Silva, D.F., Rosa, H., Carvalho, A.F.A., Oliva-Neto, P., 2015. Immobilization of pa- ica papaya papain sequence in the methylotrophic yeast Pichia pastoris. Journal of
pain on chitin and chitosan and recycling of soluble enzyme for deflocculation of Microbial & Biochemical Technology 7, 313–317.
saccharomyces cerevisiae from bioethanol distilleries. Enzyme Research 2015, Xin, B.J., Si, S.F., Xing, G.W., 2010. Protease immobilization on γ-Fe2O3/Fe3O4
1–10. magnetic nanoparticles for the synthesis of oligopeptides in organic solvents.
Subramaniam, P., Gilhotra, K., 2011. Antimicrobial efficacy of an indigenously pre-
pared caries removing gel. Contemporary Clinical Dentistry 2, 13–16.
Taylor, M.A., Pratt, K.A., Revell, D.F., Baker, K.C., Sumner, I.G., Goodenough, P.W.,
1992. Active papain renatured and processed from insoluble recombinant propa-
pain expressed in. Escherichia coli. Protein Engineering, Design and Selection 5,
TE Chemistry-an Asian Journal 5, 1389–1394.
Xue, Y., Nie, H., Zhu, L., Li, S., Zhang, H., 2010. Immobilization of modified papain
with anhydride groups on activated cotton fabric. Applied Biochemistry and
Biotechnology 160, 109–121.
You, L., Regenstein, J.M., Liu, R.H., 2010. Optimization of hydrolysis conditions for
455–459. the production of antioxidant peptides from fish gelatin using response surface
Tsuge, H., Nishimura, T., Tada, Y., Asao, T., Turk, D., Turk, V., et al., 1999. Inhibi- methodology. Journal of Food Science 75, C582–C587.
EC
tion mechanism of cathepsin L-specific inhibitors based on the crystal structure of Zhou, L., Wang, C., Jiang, Y., Gao, J., 2013. Immobilization of papain in biosilica ma-
papain–CLIK148 complex. Biochemical and Biophysical Research Communica- trix and its catalytic property. Chinese Journal of Chemical Engineering 21,
tions 266, 411–416. 670–675.
RR
CO
UN

You might also like